Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Immunity ; 53(2): 398-416.e8, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814028

RESUMEN

Paneth cells are the primary source of C-type lysozyme, a ß-1,4-N-acetylmuramoylhydrolase that enzymatically processes bacterial cell walls. Paneth cells are normally present in human cecum and ascending colon, but are rarely found in descending colon and rectum; Paneth cell metaplasia in this region and aberrant lysozyme production are hallmarks of inflammatory bowel disease (IBD) pathology. Here, we examined the impact of aberrant lysozyme production in colonic inflammation. Targeted disruption of Paneth cell lysozyme (Lyz1) protected mice from experimental colitis. Lyz1-deficiency diminished intestinal immune responses to bacterial molecular patterns and resulted in the expansion of lysozyme-sensitive mucolytic bacteria, including Ruminococcus gnavus, a Crohn's disease-associated pathobiont. Ectopic lysozyme production in colonic epithelium suppressed lysozyme-sensitive bacteria and exacerbated colitis. Transfer of R. gnavus into Lyz1-/- hosts elicited a type 2 immune response, causing epithelial reprograming and enhanced anti-colitogenic capacity. In contrast, in lysozyme-intact hosts, processed R. gnavus drove pro-inflammatory responses. Thus, Paneth cell lysozyme balances intestinal anti- and pro-inflammatory responses, with implications for IBD.


Asunto(s)
Clostridiales/inmunología , Colitis Ulcerosa/patología , Muramidasa/genética , Muramidasa/metabolismo , Células de Paneth/metabolismo , Animales , Clostridiales/genética , Colitis Ulcerosa/microbiología , Enfermedad de Crohn/patología , Femenino , Microbioma Gastrointestinal/genética , Células Caliciformes/citología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT6/genética
2.
Genes Dev ; 32(21-22): 1430-1442, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30366903

RESUMEN

After acquiring competence for selected cell fates, embryonic primordia may remain plastic for variable periods before tissue identity is irrevocably determined (commitment). We investigated the chromatin basis for these developmental milestones in mouse endoderm, a tissue with recognizable rostro-caudal patterning and transcription factor (TF)-dependent interim plasticity. Foregut-specific enhancers are as accessible and active in early midgut as in foregut endoderm, and intestinal enhancers and identity are established only after ectopic cis-regulatory elements are decommissioned. Depletion of the intestinal TF CDX2 before this cis element transition stabilizes foregut enhancers, reinforces ectopic transcriptional programs, and hence imposes foregut identities on the midgut. Later in development, as the window of chromatin plasticity elapses, CDX2 depletion weakens intestinal, without strengthening foregut, enhancers. Thus, midgut endoderm is primed for heterologous cell fates, and TFs act on a background of shifting chromatin access to determine intestinal at the expense of foregut identity. Similar principles likely govern other fate commitments.


Asunto(s)
Endodermo/metabolismo , Elementos de Facilitación Genéticos , Mucosa Intestinal/metabolismo , Intestinos/embriología , Transcripción Genética , Animales , Factor de Transcripción CDX2/genética , Factor de Transcripción CDX2/metabolismo , Cromatina/metabolismo , Endodermo/embriología , Intestinos/anatomía & histología , Ratones
3.
Development ; 149(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34910127

RESUMEN

Although Wnt signaling is clearly important for the intestinal epithelial homeostasis, the relevance of various sources of Wnt ligands themselves remains incompletely understood. Blocking the release of Wnt in distinct stromal cell types suggests obligatory functions of several stromal cell sources and yields different observations. The physiological contribution of epithelial Wnt to tissue homeostasis remains unclear. We show here that blocking epithelial Wnts affects colonic Reg4+ epithelial cell differentiation and impairs colonic epithelial regeneration after injury in mice. Single-cell RNA analysis of intestinal stroma showed that the majority of Wnt-producing cells were contained in transgelin (Tagln+) and smooth muscle actin α2 (Acta2+) expressing populations. We genetically attenuated Wnt production from these stromal cells using Tagln-Cre and Acta2-CreER drivers, and found that blockage of Wnt release from either epithelium or Tagln+ and Acta2+ stromal cells impaired colonic epithelial healing after chemical-induced injury. Aggregated blockage of Wnt release from both epithelium and Tagln+ or Acta2+ stromal cells drastically diminished epithelial repair, increasing morbidity and mortality. These results from two uncharacterized stromal populations suggested that colonic recovery from colitis-like injury depends on multiple Wnt-producing sources.


Asunto(s)
Actinas/metabolismo , Colitis Ulcerosa/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Proteína Wnt3A/metabolismo , Cicatrización de Heridas , Actinas/genética , Animales , Células Cultivadas , Colon/citología , Colon/metabolismo , Colon/fisiología , Mucosa Intestinal/citología , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/metabolismo , Proteína Wnt3A/genética
4.
EMBO Rep ; 24(9): e56240, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37424454

RESUMEN

RAB11 small GTPases and associated recycling endosome have been localized to mitotic spindles and implicated in regulating mitosis. However, the physiological significance of such regulation has not been observed in mammalian tissues. We have used newly engineered mouse models to investigate intestinal epithelial renewal in the absence of single or double isoforms of RAB11 family members: Rab11a and Rab11b. Comparing with single knockouts, mice with compound ablation demonstrate a defective cell cycle entry and robust mitotic arrest followed by apoptosis, leading to a total penetrance of lethality within 3 days of gene ablation. Upon Rab11 deletion ex vivo, enteroids show abnormal mitotic spindle formation and cell death. Untargeted proteomic profiling of Rab11a and Rab11b immunoprecipitates has uncovered a shared interactome containing mitotic spindle microtubule regulators. Disrupting Rab11 alters kinesin motor KIF11 function and impairs bipolar spindle formation and cell division. These data demonstrate that RAB11A and RAB11B redundantly control mitotic spindle function and intestinal progenitor cell division, a mechanism that may be utilized to govern the homeostasis and renewal of other mammalian tissues.


Asunto(s)
Proteómica , Proteínas de Unión al GTP rab , Animales , Ratones , Mamíferos/metabolismo , Mitosis , Proteínas de Unión al GTP rab/metabolismo , Huso Acromático/metabolismo , Células Madre/metabolismo
5.
Brain Behav Immun ; 119: 665-680, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38579936

RESUMEN

Depression is a prevalent psychological condition with limited treatment options. While its etiology is multifactorial, both chronic stress and changes in microbiome composition are associated with disease pathology. Stress is known to induce microbiome dysbiosis, defined here as a change in microbial composition associated with a pathological condition. This state of dysbiosis is known to feedback on depressive symptoms. While studies have demonstrated that targeted restoration of the microbiome can alleviate depressive-like symptoms in mice, translating these findings to human patients has proven challenging due to the complexity of the human microbiome. As such, there is an urgent need to identify factors upstream of microbial dysbiosis. Here we investigate the role of mucin 13 as an upstream mediator of microbiome composition changes in the context of stress. Using a model of chronic stress, we show that the glycocalyx protein, mucin 13, is selectively reduced after psychological stress exposure. We further demonstrate that the reduction of Muc13 is mediated by the Hnf4 transcription factor family. Finally, we determine that deleting Muc13 is sufficient to drive microbiome shifts and despair behaviors. These findings shed light on the mechanisms behind stress-induced microbial changes and reveal a novel regulator of mucin 13 expression.


Asunto(s)
Depresión , Disbiosis , Microbioma Gastrointestinal , Estrés Psicológico , Animales , Masculino , Ratones , Conducta Animal/fisiología , Depresión/metabolismo , Depresión/microbiología , Disbiosis/metabolismo , Disbiosis/microbiología , Microbioma Gastrointestinal/fisiología , Factor Nuclear 4 del Hepatocito/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mucinas/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/microbiología
6.
J Biol Chem ; 298(8): 102213, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35779631

RESUMEN

1,25-dihydroxyvitamin D (VD) regulates intestinal calcium absorption in the small intestine (SI) and also reduces risk of colonic inflammation and cancer. However, the intestine compartment-specific target genes of VD signaling are unknown. Here, we examined VD action across three functional compartments of the intestine using RNA-seq to measure VD-induced changes in gene expression and Chromatin Immunoprecipitation with next generation sequencing to measure vitamin D receptor (VDR) genomic binding. We found that VD regulated the expression of 55 shared transcripts in the SI crypt, SI villi, and in the colon, including Cyp24a1, S100g, Trpv6, and Slc30a10. Other VD-regulated transcripts were unique to the SI crypt (162 up, 210 down), villi (199 up, 63 down), or colon (102 up, 28 down), but this did not correlate with mRNA levels of the VDR. Furthermore, bioinformatic analysis identified unique VD-regulated biological functions in each compartment. VDR-binding sites were found in 70% of upregulated genes from the colon and SI villi but were less common in upregulated genes from the SI crypt and among downregulated genes, suggesting some transcript-level VD effects are likely indirect. Consistent with this, we show that VD regulated the expression of other transcription factors and their downstream targets. Finally, we demonstrate that compartment-specific VD-mediated gene expression was associated with compartment-specific VDR-binding sites (<30% of targets) and enrichment of intestinal transcription factor-binding motifs within VDR-binding peaks. Taken together, our data reveal unique spatial patterns of VD action in the intestine and suggest novel mechanisms that could account for compartment-specific functions of this hormone.


Asunto(s)
Receptores de Calcitriol , Vitamina D , Animales , Genómica , Intestinos , Ratones , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Vitamina D/farmacología , Vitamina D3 24-Hidroxilasa/genética
7.
Development ; 146(19)2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31345929

RESUMEN

As embryos mature, cells undergo remarkable transitions that are accompanied by shifts in transcription factor regulatory networks. Mechanisms driving developmental transitions are incompletely understood. The embryonic intestine transitions from a rapidly proliferating tube with pseudostratified epithelium prior to murine embryonic day (E) 14.5 to an exquisitely folded columnar epithelium in fetal stages. We sought to identify factors driving mouse fetal intestinal maturation by mining chromatin accessibility data for transcription factor motifs. ATAC-seq accessible regions shift during tissue maturation, with CDX2 transcription factor motifs abundant at chromatin-accessible regions of the embryo. Hepatocyte nuclear factor 4 (HNF4) transcription factor motifs are the most abundant in the fetal stages (>E16.5). Genetic inactivation of Hnf4a and its paralog Hnf4g revealed that HNF4 factors are redundantly required for fetal maturation. CDX2 binds to and activates Hnf4 gene loci to elevate HNF4 expression at fetal stages. HNF4 and CDX2 transcription factors then occupy shared genomic regulatory sites to promote chromatin accessibility and gene expression in the maturing intestine. Thus, HNF4 paralogs are key components of an intestinal transcription factor network shift during the embryonic to fetal transition.


Asunto(s)
Cromatina/metabolismo , Feto/embriología , Factor Nuclear 4 del Hepatocito/metabolismo , Intestinos/embriología , Animales , Factor de Transcripción CDX2/metabolismo , Línea Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Modelos Biológicos , Morfogénesis
8.
Development ; 146(5)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30745430

RESUMEN

Lineage-restricted transcription factors, such as the intestine-specifying factor CDX2, often have dual requirements across developmental time. Embryonic loss of CDX2 triggers homeotic transformation of intestinal fate, whereas adult-onset loss compromises crucial physiological functions but preserves intestinal identity. It is unclear how such diverse requirements are executed across the developmental continuum. Using primary and engineered human tissues, mouse genetics, and a multi-omics approach, we demonstrate that divergent CDX2 loss-of-function phenotypes in embryonic versus adult intestines correspond to divergent CDX2 chromatin-binding profiles in embryonic versus adult stages. CDX2 binds and activates distinct target genes in developing versus adult mouse and human intestinal cells. We find that temporal shifts in chromatin accessibility correspond to these context-specific CDX2 activities. Thus, CDX2 is not sufficient to activate a mature intestinal program; rather, CDX2 responds to its environment, targeting stage-specific genes to contribute to either intestinal patterning or mature intestinal function. This study provides insights into the mechanisms through which lineage-specific regulatory factors achieve divergent functions over developmental time.


Asunto(s)
Factor de Transcripción CDX2/metabolismo , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Intestinos/embriología , Animales , Factor de Transcripción CDX2/genética , Sistemas CRISPR-Cas , Diferenciación Celular , Linaje de la Célula , Femenino , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Mutación , Células Madre Pluripotentes/citología , Unión Proteica , Dominios Proteicos , Transactivadores/metabolismo
9.
Physiol Genomics ; 53(11): 486-508, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34612061

RESUMEN

Human intestinal epithelial organoids (enteroids and colonoids) are tissue cultures used for understanding the physiology of the human intestinal epithelium. Here, we explored the effect on the transcriptome of common variations in culture methods, including extracellular matrix substrate, format, tissue segment, differentiation status, and patient heterogeneity. RNA-sequencing datasets from 276 experiments performed on 37 human enteroid and colonoid lines from 29 patients were aggregated from several groups in the Texas Medical Center. DESeq2 and gene set enrichment analysis (GSEA) were used to identify differentially expressed genes and enriched pathways. PERMANOVA, Pearson's correlation, and dendrogram analysis of the data originally indicated three tiers of influence of culture methods on transcriptomic variation: substrate (collagen vs. Matrigel) and format (3-D, transwell, and monolayer) had the largest effect; segment of origin (duodenum, jejunum, ileum, colon) and differentiation status had a moderate effect; and patient heterogeneity and specific experimental manipulations (e.g., pathogen infection) had the smallest effect. GSEA identified hundreds of pathways that varied between culture methods, such as IL1 cytokine signaling enriched in transwell versus monolayer cultures and E2F target genes enriched in collagen versus Matrigel cultures. The transcriptional influence of the format was furthermore validated in a synchronized experiment performed with various format-substrate combinations. Surprisingly, large differences in organoid transcriptome were driven by variations in culture methods such as format, whereas experimental manipulations such as infection had modest effects. These results show that common variations in culture conditions can have large effects on intestinal organoids and should be accounted for when designing experiments and comparing results between laboratories. Our data constitute the largest RNA-seq dataset interrogating human intestinal epithelial organoids.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Colon/metabolismo , Medios de Cultivo/farmacología , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Organoides/metabolismo , Transcriptoma/efectos de los fármacos , Calcitriol/farmacología , Colágeno/metabolismo , Colágeno/farmacología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Medios de Cultivo/química , Combinación de Medicamentos , Escherichia coli , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Laminina/metabolismo , Laminina/farmacología , Organoides/virología , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , RNA-Seq/métodos , Transcriptoma/genética , Virosis/metabolismo , Virosis/virología , Virus
10.
Gastroenterology ; 158(4): 985-999.e9, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31759926

RESUMEN

BACKGROUND & AIMS: Functions of intestinal stem cells (ISCs) are regulated by diet and metabolic pathways. Hepatocyte nuclear factor 4 (HNF4) family are transcription factors that bind fatty acids. We investigated how HNF4 transcription factors regulate metabolism and their functions in ISCs in mice. METHODS: We performed studies with Villin-CreERT2;Lgr5-EGFP-IRES-CreERT2;Hnf4αf/f;Hnf4γCrispr/Crispr mice, hereafter referred to Hnf4αγDKO. Mice were given tamoxifen to induce Cre recombinase. Mice transgenic with only Cre alleles (Villin-CreERT2, Lgr5-EGFP-IRES-CreERT2, Hnf4α+/+, and Hnf4γ+/+) or mice given vehicle were used as controls. Crypt and villus cells were isolated, incubated with fluorescently labeled fatty acids or glucose analog, and analyzed by confocal microscopy. Fatty acid oxidation activity and tricarboxylic acid (TCA) cycle metabolites were measured in cells collected from the proximal half of the small intestine of Hnf4αγDKO and control mice. We performed chromatin immunoprecipitation and gene expression profiling analyses to identify genes regulated by HNF4 factors. We established organoids from duodenal crypts, incubated them with labeled palmitate or acetate, and measured production of TCA cycle metabolites or fatty acids. Acetate, a precursor of acetyl coenzyme A (CoA) (a product of fatty acid ß-oxidation [FAO]), or dichloroacetate, a compound that promotes pyruvate oxidation and generation of mitochondrial acetyl-CoA, were used for metabolic intervention. RESULTS: Crypt cells rapidly absorbed labeled fatty acids, and messenger RNA levels of Lgr5+ stem cell markers (Lgr5, Olfm4, Smoc2, Msi1, and Ascl2) were down-regulated in organoids incubated with etomoxir, an inhibitor of FAO, indicating that FAO was required for renewal of ISCs. HNF4A and HNF4G were expressed in ISCs and throughout the intestinal epithelium. Single knockout of either HNF4A or HNF4G did not affect maintenance of ISCs, but double-knockout of HNF4A and HNF4G resulted in ISC loss; stem cells failed to renew. FAO supports ISC renewal, and HNF4 transcription factors directly activate FAO genes, including Acsl5 and Acsf2 (encode regulators of acyl-CoA synthesis), Slc27a2 (encodes a fatty acid transporter), Fabp2 (encodes fatty acid binding protein), and Hadh (encodes hydroxyacyl-CoA dehydrogenase). In the intestinal epithelium of Hnf4αγDKO mice, expression levels of FAO genes, FAO activity, and metabolites of TCA cycle were all significantly decreased, but fatty acid synthesis transcripts were increased, compared with control mice. The contribution of labeled palmitate or acetate to the TCA cycle was reduced in organoids derived from Hnf4αγDKO mice, compared with control mice. Incubation of organoids derived from double-knockout mice with acetate or dichloroacetate restored stem cells. CONCLUSIONS: In mice, the transcription factors HNF4A and HNF4G regulate the expression of genes required for FAO and are required for renewal of ISCs.


Asunto(s)
Ácidos Grasos/metabolismo , Factor Nuclear 4 del Hepatocito/fisiología , Intestino Delgado/citología , Células Madre/metabolismo , Animales , Duodeno/citología , Proteínas de Unión a Ácidos Grasos/metabolismo , Mucosa Intestinal/citología , Ratones , Ratones Noqueados , Organoides/metabolismo , Oxidación-Reducción
11.
Nucleic Acids Res ; 47(15): 7870-7885, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31226208

RESUMEN

Long interspersed elements-1 (LINE-1, L1) are retrotransposons that hold the capacity of self-propagation in the genome with potential mutagenic outcomes. How somatic cells restrict L1 activity and how this process becomes dysfunctional during aging and in cancer cells is poorly understood. L1s are enriched at lamin-associated domains, heterochromatic regions of the nuclear periphery. Whether this association is necessary for their repression has been elusive. Here we show that the sirtuin family member SIRT7 participates in the epigenetic transcriptional repression of L1 genome-wide in both mouse and human cells. SIRT7 depletion leads to increased L1 expression and retrotransposition. Mechanistically, we identify a novel interplay between SIRT7 and Lamin A/C in L1 repression. Our results demonstrate that SIRT7-mediated H3K18 deacetylation regulates L1 expression and promotes L1 association with elements of the nuclear lamina. The failure of such activity might contribute to the observed genome instability and compromised viability in SIRT7 knockout mice. Overall, our results reveal a novel function of SIRT7 on chromatin organization by mediating the anchoring of L1 to the nuclear envelope, and a new functional link of the nuclear lamina with transcriptional repression.


Asunto(s)
Genoma , Lamina Tipo A/genética , Elementos de Nucleótido Esparcido Largo , Sirtuinas/genética , Transcripción Genética , Animales , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/metabolismo , Heterocromatina/química , Heterocromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Células K562 , Lamina Tipo A/metabolismo , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocardio/citología , Miocardio/metabolismo , Lámina Nuclear/metabolismo , Lámina Nuclear/ultraestructura , Sirtuinas/deficiencia , Sirtuinas/metabolismo , Testículo/citología , Testículo/metabolismo
12.
J Biol Chem ; 294(11): 3974-3986, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30659096

RESUMEN

Hypoxia-inducible factor 2α (HIF2α) directly regulates a battery of genes essential for intestinal iron absorption. Interestingly, iron deficiency and overload disorders do not result in increased intestinal expression of glycolytic or angiogenic HIF2α target genes. Similarly, inflammatory and tumor foci can induce a distinct subset of HIF2α target genes in vivo These observations indicate that different stimuli activate distinct subsets of HIF2α target genes via mechanisms that remain unclear. Here, we conducted a high-throughput siRNA-based screen to identify genes that regulate HIF2α's transcriptional activity on the promoter of the iron transporter gene divalent metal transporter-1 (DMT1). SMAD family member 3 (SMAD3) and SMAD4 were identified as potential transcriptional repressors. Further analysis revealed that SMAD4 signaling selectively represses iron-absorptive gene promoters but not the inflammatory or glycolytic HIF2α or HIF1α target genes. Moreover, the highly homologous SMAD2 did not alter HIF2α transcriptional activity. During iron deficiency, SMAD3 and SMAD4 expression was significantly decreased via proteasomal degradation, allowing for derepression of iron target genes. Several iron-regulatory genes contain a SMAD-binding element (SBE) in their proximal promoters; however, mutation of the putative SBE on the DMT1 promoter did not alter the repressive function of SMAD3 or SMAD4. Importantly, the transcription factor forkhead box protein A1 (FOXA1) was critical in SMAD4-induced DMT1 repression, and DNA binding of SMAD4 was essential for the repression of HIF2α activity, suggesting an indirect repressive mechanism through DNA binding. These results provide mechanistic clues to how HIF signaling can be regulated by different cellular cues.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Proteína smad3/metabolismo , Proteína Smad4/metabolismo , Animales , Células Cultivadas , Humanos , Proteínas Reguladoras del Hierro/genética , Ratones , Ratones Noqueados , Proteína smad3/deficiencia , Proteína Smad4/deficiencia
13.
Am J Physiol Gastrointest Liver Physiol ; 319(2): G189-G196, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32628072

RESUMEN

To fulfill the lifelong need to supply diverse epithelial cells, intestinal stem cells (ISCs) rely on executing accurate transcriptional programs. This review addresses the mechanisms that control those programs. Genes that define cell behaviors and identities are regulated principally through thousands of dispersed enhancers, each individually <1 kb long and positioned from a few to hundreds of kilobases away from transcription start sites, upstream or downstream from coding genes or within introns. Wnt, Notch, and other epithelial control signals feed into these cis-regulatory DNA elements, which are also common loci of polymorphisms and mutations that confer disease risk. Cell-specific gene activity requires promoters to interact with the correct combination of signal-responsive enhancers. We review the current state of knowledge in ISCs regarding active enhancers, the nucleosome modifications that may enable appropriate and hinder inappropriate enhancer-promoter contacts, and the roles of lineage-restricted transcription factors.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética , Intestinos/citología , Células Madre/fisiología , Animales , Regulación de la Expresión Génica , Humanos
14.
Dev Biol ; 439(2): 92-101, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29684311

RESUMEN

During development, the embryo transitions from a metabolism favoring glycolysis to a metabolism favoring mitochondrial respiration. How metabolic shifts regulate developmental processes, or how developmental processes regulate metabolic shifts, remains unclear. To test the requirement of mitochondrial function in developing endoderm-derived tissues, we genetically inactivated the mitochondrial transcription factor, Tfam, using the Shh-Cre driver. Tfam mutants did not survive postnatally, exhibiting defects in lung development. In the developing intestine, TFAM-loss was tolerated until late fetal development, during which the process of villus elongation was compromised. While progenitor cell populations appeared unperturbed, markers of enterocyte maturation were diminished and villi were blunted. Loss of TFAM was also tested in the adult intestinal epithelium, where enterocyte maturation was similarly dependent upon the mitochondrial transcription factor. While progenitor cells in the transit amplifying zone of the adult intestine remained proliferative, intestinal stem cell renewal was dependent upon TFAM, as indicated by molecular profiling and intestinal organoid formation assays. Taken together, these studies point to critical roles for the mitochondrial regulator TFAM for multiple aspects of intestinal development and maturation, and highlight the importance of mitochondrial regulators in tissue development and homeostasis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Grupo de Alta Movilidad/fisiología , Mucosa Intestinal/metabolismo , Animales , Técnicas de Cultivo de Célula/métodos , Autorrenovación de las Células/fisiología , Proteínas de Unión al ADN/genética , Feto/metabolismo , Regulación de la Expresión Génica/genética , Glucólisis/genética , Glucólisis/fisiología , Proteínas del Grupo de Alta Movilidad/genética , Mucosa Intestinal/embriología , Mucosa Intestinal/crecimiento & desarrollo , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Organogénesis/genética , Organogénesis/fisiología , Organoides/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo
15.
Development ; 143(20): 3711-3722, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27802136

RESUMEN

During late gestation, villi extend into the intestinal lumen to dramatically increase the surface area of the intestinal epithelium, preparing the gut for the neonatal diet. Incomplete development of the intestine is the most common gastrointestinal complication in neonates, but the causes are unclear. We provide evidence in mice that Yin Yang 1 (Yy1) is crucial for intestinal villus development. YY1 loss in the developing endoderm had no apparent consequences until late gestation, after which the intestine differentiated poorly and exhibited severely stunted villi. Transcriptome analysis revealed that YY1 is required for mitochondrial gene expression, and ultrastructural analysis confirmed compromised mitochondrial integrity in the mutant intestine. We found increased oxidative phosphorylation gene expression at the onset of villus elongation, suggesting that aerobic respiration might function as a regulator of villus growth. Mitochondrial inhibitors blocked villus growth in a fashion similar to Yy1 loss, thus further linking oxidative phosphorylation with late-gestation intestinal development. Interestingly, we find that necrotizing enterocolitis patients also exhibit decreased expression of oxidative phosphorylation genes. Our study highlights the still unappreciated role of metabolic regulation during organogenesis, and suggests that it might contribute to neonatal gastrointestinal disorders.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/citología , Organogénesis/fisiología , Factor de Transcripción YY1/metabolismo , Aerobiosis/genética , Aerobiosis/fisiología , Animales , Western Blotting , Genotipo , Inmunohistoquímica , Masculino , Ratones , Organogénesis/genética , Fosforilación Oxidativa , Transcriptoma/genética , Factor de Transcripción YY1/genética
16.
Carcinogenesis ; 39(5): 669-680, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29547900

RESUMEN

Inflammation is highly associated with colon carcinogenesis. Epigenetic mechanisms could play an important role in the initiation and progression of colon cancer. Curcumin, a dietary phytochemical, shows promising effects in suppressing colitis-associated colon cancer in azoxymethane-dextran sulfate sodium (AOM-DSS) mice. However, the potential epigenetic mechanisms of curcumin in colon cancer remain unknown. In this study, the anticancer effect of curcumin in suppressing colon cancer in an 18-week AOM-DSS colon cancer mouse model was confirmed. We identified lists of differentially expressed and differentially methylated genes in pairwise comparisons and several pathways involved in the potential anticancer effect of curcumin. These pathways include LPS/IL-1-mediated inhibition of RXR function, Nrf2-mediated oxidative stress response, production of NO and ROS in macrophages and IL-6 signaling. Among these genes, Tnf stood out with decreased DNA CpG methylation of Tnf in the AOM-DSS group and reversal of the AOM-DSS induced Tnf demethylation by curcumin. These observations in Tnf methylation correlated with increased and decreased Tnf expression in RNA-seq. The functional role of DNA methylation of Tnf was further confirmed by in vitro luciferase transcriptional activity assay. In addition, the DNA methylation level in a group of inflammatory genes was decreased in the AOM+DSS group but restored by curcumin and was validated by pyrosequencing. This study shows for the first time epigenomic changes in DNA CpG methylation in the inflammatory response from colitis-associated colon cancer and the reversal of their CpG methylation changes by curcumin. Future clinical epigenetic studies with curcumin in inflammation-associated colon cancer would be warranted.


Asunto(s)
Colitis/complicaciones , Neoplasias del Colon/etiología , Neoplasias del Colon/prevención & control , Curcumina/farmacología , Metilación de ADN/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Animales , Azoximetano/farmacología , Colon/efectos de los fármacos , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos
17.
Development ; 142(16): 2775-80, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26160899

RESUMEN

Endothelin signaling is essential for neural crest development, and dysregulated Endothelin signaling is associated with several neural crest-related disorders, including Waardenburg and other syndromes. However, despite the crucial roles of this pathway in neural crest development and disease, the transcriptional effectors directly activated by Endothelin signaling during neural crest development remain incompletely elucidated. Here, we establish that the MADS box transcription factor MEF2C is an immediate downstream transcriptional target and effector of Endothelin signaling in the neural crest. We show that Endothelin signaling activates Mef2c expression in the neural crest through a conserved enhancer in the Mef2c locus and that CRISPR-mediated deletion of this Mef2c neural crest enhancer from the mouse genome abolishes Endothelin induction of Mef2c expression. Moreover, we demonstrate that Endothelin signaling activates neural crest expression of Mef2c by de-repressing MEF2C activity through a Calmodulin-CamKII-histone deacetylase signaling cascade. Thus, these findings identify a MEF2C-dependent, positive-feedback mechanism for Endothelin induction and establish MEF2C as an immediate transcriptional effector and target of Endothelin signaling in the neural crest.


Asunto(s)
Endotelinas/metabolismo , Retroalimentación Fisiológica/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Cresta Neural/fisiología , Transducción de Señal/fisiología , Animales , Galactósidos , Hibridación in Situ , Indoles , Factores de Transcripción MEF2/metabolismo , Ratones , Ratones Transgénicos , Cresta Neural/metabolismo , beta-Galactosidasa
18.
Development ; 142(12): 2147-62, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26015543

RESUMEN

Communication between stem and niche supporting cells maintains the homeostasis of adult tissues. Wnt signaling is a crucial regulator of the stem cell niche, but the mechanism that governs Wnt ligand delivery in this compartment has not been fully investigated. We identified that Wnt secretion is partly dependent on Rab8a-mediated anterograde transport of Gpr177 (wntless), a Wnt-specific transmembrane transporter. Gpr177 binds to Rab8a, depletion of which compromises Gpr177 traffic, thereby weakening the secretion of multiple Wnts. Analyses of generic Wnt/ß-catenin targets in Rab8a knockout mouse intestinal crypts indicate reduced signaling activities; maturation of Paneth cells - a Wnt-dependent cell type - is severely affected. Rab8a knockout crypts show an expansion of Lgr5(+) and Hopx(+) cells in vivo. However, in vitro, the knockout enteroids exhibit significantly weakened growth that can be partly restored by exogenous Wnts or Gsk3ß inhibitors. Immunogold labeling and surface protein isolation identified decreased plasma membrane localization of Gpr177 in Rab8a knockout Paneth cells and fibroblasts. Upon stimulation by exogenous Wnts, Rab8a-deficient cells show ligand-induced Lrp6 phosphorylation and transcriptional reporter activation. Rab8a thus controls Wnt delivery in producing cells and is crucial for Paneth cell maturation. Our data highlight the profound tissue plasticity that occurs in response to stress induced by depletion of a stem cell niche signal.


Asunto(s)
Células de Paneth/citología , Nicho de Células Madre/fisiología , Células Madre/citología , Proteínas de Unión al GTP rab/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo , Activación Transcripcional , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Proteínas de Unión al GTP rab/genética
19.
Proc Natl Acad Sci U S A ; 111(21): 7695-700, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24821761

RESUMEN

The intestinal stem cell fuels the highest rate of tissue turnover in the body and has been implicated in intestinal disease and cancer; understanding the regulatory mechanisms controlling intestinal stem cell physiology is of great importance. Here, we provide evidence that the transcription factor YY1 is essential for intestinal stem cell renewal. We observe that YY1 loss skews normal homeostatic cell turnover, with an increase in proliferating crypt cells and a decrease in their differentiated villous progeny. Increased crypt cell numbers come at the expense of Lgr5(+) stem cells. On YY1 deletion, Lgr5(+) cells accelerate their commitment to the differentiated population, exhibit increased levels of apoptosis, and fail to maintain stem cell renewal. Loss of Yy1 in the intestine is ultimately fatal. Mechanistically, YY1 seems to play a role in stem cell energy metabolism, with mitochondrial complex I genes bound directly by YY1 and their transcript levels decreasing on YY1 loss. These unappreciated YY1 functions broaden our understanding of metabolic regulation in intestinal stem cell homeostasis.


Asunto(s)
División Celular/fisiología , Regulación de la Expresión Génica/fisiología , Intestinos/citología , Mitocondrias/metabolismo , Células Madre/fisiología , Factor de Transcripción YY1/metabolismo , Animales , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Análisis por Micromatrices , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Factor de Transcripción YY1/genética
20.
J Biol Chem ; 290(3): 1850-60, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25488664

RESUMEN

Distinct groups of transcription factors (TFs) assemble at tissue-specific cis-regulatory sites, implying that different TF combinations may control different genes and cellular functions. Within such combinations, TFs that specify or maintain a lineage and are therefore considered master regulators may play a key role. Gene enhancers often attract these tissue-restricted TFs, as well as TFs that are expressed more broadly. However, the contributions of the individual TFs to combinatorial regulatory activity have not been examined critically in many cases in vivo. We address this question using a genetic approach in mice to inactivate the intestine-specifying and intestine-restricted factor CDX2 alone or in combination with its more broadly expressed partner factors, GATA4 and HNF4A. Compared with single mutants, each combination produced significantly greater defects and rapid lethality through distinct anomalies. Intestines lacking Gata4 and Cdx2 were deficient in crypt cell replication, whereas combined loss of Hnf4a and Cdx2 specifically impaired viability and maturation of villus enterocytes. Integrated analysis of TF binding and of transcripts affected in Hnf4a;Cdx2 compound-mutant intestines indicated that this TF pair controls genes required to construct the apical brush border and absorb nutrients, including dietary lipids. This study thus defines combinatorial TF activities, their specific requirements during tissue homeostasis, and modules of transcriptional targets in intestinal epithelial cells in vivo.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Mucosa Intestinal/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Factor de Transcripción CDX2 , Diferenciación Celular , Inmunoprecipitación de Cromatina , Enterocitos/citología , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda